Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nat Immunol ; 13(3): 223-228, 2012 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-22327569

RESUMEN

SAMHD1 restricts the infection of dendritic and other myeloid cells by human immunodeficiency virus type 1 (HIV-1), but in lentiviruses of the simian immunodeficiency virus of sooty mangabey (SIVsm)-HIV-2 lineage, SAMHD1 is counteracted by the virion-packaged accessory protein Vpx. Here we found that SAMHD1 restricted infection by hydrolyzing intracellular deoxynucleoside triphosphates (dNTPs), lowering their concentrations to below those required for the synthesis of the viral DNA by reverse transcriptase (RT). SAMHD1-mediated restriction was alleviated by the addition of exogenous deoxynucleosides. An HIV-1 with a mutant RT with low affinity for dNTPs was particularly sensitive to SAMHD1-mediated restriction. Vpx prevented the SAMHD1-mediated decrease in dNTP concentration and induced the degradation of human and rhesus macaque SAMHD1 but had no effect on mouse SAMHD1. Nucleotide-pool depletion could be a general mechanism for protecting cells from infectious agents that replicate through a DNA intermediate.


Asunto(s)
VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleótidos/metabolismo , Replicación Viral , Animales , Línea Celular , Humanos , Espacio Intracelular/metabolismo , Macaca mulatta , Macrófagos/inmunología , Ratones , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/inmunología , Proteína 1 que Contiene Dominios SAM y HD
2.
Proc Natl Acad Sci U S A ; 113(19): 5311-6, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27114546

RESUMEN

Viruses often interfere with the DNA damage response to better replicate in their hosts. The human immunodeficiency virus 1 (HIV-1) viral protein R (Vpr) protein has been reported to modulate the activity of the DNA repair structure-specific endonuclease subunit (SLX4) complex and to promote cell cycle arrest. Vpr also interferes with the base-excision repair pathway by antagonizing the uracil DNA glycosylase (Ung2) enzyme. Using an unbiased quantitative proteomic screen, we report that Vpr down-regulates helicase-like transcription factor (HLTF), a DNA translocase involved in the repair of damaged replication forks. Vpr subverts the DDB1-cullin4-associated-factor 1 (DCAF1) adaptor of the Cul4A ubiquitin ligase to trigger proteasomal degradation of HLTF. This event takes place rapidly after Vpr delivery to cells, before and independently of Vpr-mediated G2 arrest. HLTF is degraded in lymphocytic cells and macrophages infected with Vpr-expressing HIV-1. Our results reveal a previously unidentified strategy for HIV-1 to antagonize DNA repair in host cells.


Asunto(s)
Daño del ADN/fisiología , Reparación del ADN/fisiología , Proteínas de Unión al ADN/metabolismo , Macrófagos/metabolismo , Linfocitos T/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Células HeLa , Humanos , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana
3.
J Mammary Gland Biol Neoplasia ; 21(1-2): 1-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27048289

RESUMEN

In the mammary glands of lactating animals, the mammary epithelial cells that surround the lumen of the acini produce and secrete copious amounts of milk. Functional differentiation of these mammary epithelial cells depends on the development of high-efficiency secretory pathways, notably for protein and lipid secretion. Protein secretion is a fundamental process common to all animal cells that involves a subset of cellular organelles, including the endoplasmic reticulum and the Golgi apparatus. In contrast, en masse secretion of triglycerides and cholesterol esters in the form of milk fat globules is a unique feature of the mammary epithelial cell. Cytoplasmic lipid droplets, the intracellular precursors of milk fat globules, originate from the endoplasmic reticulum, as do most milk-specific proteins. This organelle is therefore pivotal in the biogenesis of milk components. Fractionation of the cell into its subcellular parts is an approach that has proven very powerful for understanding organelle function and for studying the specific role of an organelle in a given cell activity. Here we describe a method for the purification of both smooth and rough microsomes, the membrane-bound endoplasmic reticulum fragments that form from endoplasmic reticulum domains when cells are broken up, from mammary gland tissue at lactation.


Asunto(s)
Retículo Endoplásmico Rugoso/metabolismo , Retículo Endoplásmico Liso/metabolismo , Lactancia/metabolismo , Glándulas Mamarias Animales/metabolismo , Animales , Biomarcadores/metabolismo , Fraccionamiento Celular , Centrifugación por Gradiente de Densidad , Retículo Endoplásmico Rugoso/ultraestructura , Retículo Endoplásmico Liso/ultraestructura , Epitelio/metabolismo , Epitelio/ultraestructura , Femenino , Cabras , Glándulas Mamarias Animales/ultraestructura , Microscopía Electrónica de Transmisión , Microsomas/metabolismo , Microsomas/ultraestructura , Ratas , Especificidad de la Especie , Factores de Tiempo
4.
Proc Natl Acad Sci U S A ; 110(42): E3997-4006, 2013 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24082141

RESUMEN

Macrophages are a major target cell for HIV-1, and their infection contributes to HIV pathogenesis. We have previously shown that the cyclin-dependent kinase inhibitor p21 inhibits the replication of HIV-1 and other primate lentiviruses in human monocyte-derived macrophages by impairing reverse transcription of the viral genome. In the attempt to understand the p21-mediated restriction mechanisms, we found that p21 impairs HIV-1 and simian immunodeficiency virus (SIV)mac reverse transcription in macrophages by reducing the intracellular deoxyribonucleotide (dNTP) pool to levels below those required for viral cDNA synthesis by a SAM domain and HD domain-containing protein 1 (SAMHD1)-independent pathway. We found that p21 blocks dNTP biosynthesis by down-regulating the expression of the RNR2 subunit of ribonucleotide reductase, an enzyme essential for the reduction of ribonucleotides to dNTP. p21 inhibits RNR2 transcription by repressing E2F1 transcription factor, its transcriptional activator. Our findings unravel a cellular pathway that restricts HIV-1 and other primate lentiviruses by affecting dNTP synthesis, thereby pointing to new potential cellular targets for anti-HIV therapeutic strategies.


Asunto(s)
Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Desoxirribonucleótidos/biosíntesis , Regulación Enzimológica de la Expresión Génica , Infecciones por VIH/metabolismo , VIH-1/fisiología , Macrófagos/metabolismo , Ribonucleótido Reductasas/biosíntesis , Replicación Viral/fisiología , Células Cultivadas , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , ADN Complementario/biosíntesis , ADN Complementario/genética , ADN Viral/biosíntesis , ADN Viral/genética , Desoxirribonucleótidos/genética , Regulación hacia Abajo/genética , Factor de Transcripción E2F1/genética , Factor de Transcripción E2F1/metabolismo , Infecciones por VIH/terapia , Infecciones por VIH/virología , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/genética , Proteínas de Unión al GTP Monoméricas/metabolismo , Ribonucleótido Reductasas/genética , Proteína 1 que Contiene Dominios SAM y HD , Virus de la Inmunodeficiencia de los Simios/fisiología , Transcripción Genética/genética
5.
Virol J ; 12: 201, 2015 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-26606981

RESUMEN

BACKGROUND: SAMHD1 counteracts HIV-1 or HIV-2/SIVsmm that lacks Vpx by depleting the intracellular pool of nucleotides in myeloid cells and CD4+ quiescent T cells, thereby inhibiting the synthesis of retroviral DNA by reverse transcriptase. Depletion of nucleotides has been shown to underline the establishment of quiescence in certain cellular systems. These observations led us to investigate whether SAMHD1 could control the transition between proliferation and quiescence using the THP-1 cell model. FINDINGS: The entry of dividing THP-1 myeloid cells into a non-dividing differentiated state was monitored after addition of phorbol-12-myristate-13-acetate (PMA), an inducer of differentiation. Under PMA treatment, cells overexpressing SAMHD1 display stronger and faster adhesion to their support, compared to cells expressing a catalytically inactive form of SAMHD1, or cells depleted of SAMHD1, which appear less differentiated. After PMA removal, cells overexpressing SAMHD1 maintain low levels of cyclin A, in contrast to other cell lines. Interestingly, SAMHD1 overexpression slightly increases cell adhesion even in the absence of the differentiation inducer PMA. Finally, we found that levels of SAMHD1 are reduced in proliferating primary CD4+ T cells after T cell receptor activation, suggesting that SAMHD1 may also be involved in the transition from a quiescent state to a dividing state in primary T cells. CONCLUSIONS: Altogether, we provide evidence that SAMHD1 may facilitate some aspects of THP-1 cell differentiation. Restriction of HIV-1 by SAMHD1 may rely upon its ability to modify cell cycle parameters, in addition to the direct inhibition of reverse transcription.


Asunto(s)
Diferenciación Celular , Proliferación Celular , Monocitos/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/virología , Adhesión Celular/efectos de los fármacos , Línea Celular , VIH-1/inmunología , VIH-1/fisiología , Humanos , Monocitos/efectos de los fármacos , Proteína 1 que Contiene Dominios SAM y HD , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/metabolismo , Replicación Viral
6.
Retrovirology ; 11: 117, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25496524

RESUMEN

Vpr is one of the most enigmatic viral auxiliary proteins of HIV. During the past twenty years, several activities have been ascribed to this viral protein, but one, its ability to mediate cell cycle arrest at the G2 to M transition has been the most extensively studied. Nonetheless, the genuine role of Vpr and its pathophysiological relevance in the viral life cycle have remained mysterious. Recent work by Laguette et al. (Cell 156:134-145, 2014) provides important insight into the molecular mechanism of Vpr-mediated G2 arrest. This study highlights for the first time how Vpr recruits the SLX4 endonuclease complex and how Vpr-induced inappropriate activation of this complex leads to G2 arrest. Here, we will discuss these findings in the light of previous work to show how they change the view of Vpr's mechanism of action. We will also discuss how these findings open new questions towards the understanding of the biological function of Vpr regarding innate immune sensing.


Asunto(s)
Puntos de Control del Ciclo Celular , VIH-1/fisiología , Interacciones Huésped-Patógeno , Recombinasas/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos
7.
Retrovirology ; 10: 30, 2013 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-23497353

RESUMEN

BACKGROUND: Interferon-α (IFN-α) is an essential mediator of the antiviral response, which potently inhibits both early and late phases of HIV replication. The SAMHD1 deoxynucleoside triphosphate (dNTP) hydrolase represents the prototype of a new antiviral strategy we referred to as "nucleotide depletion". SAMHD1 depletes dNTP levels in myeloid cells below those required for optimal synthesis of HIV viral DNA. HIV-2 and its SIVsm and SIVmac close relatives encode a protein termed Vpx, which counteracts SAMHD1. The potentiality of IFN-α to cooperate with nucleotide depletion has been poorly investigated so far. Here we wondered whether IFN-α affects SAMHD1 expression, Vpx-induced SAMHD1 degradation, Vpx-mediated rescue of HIV-1 transduction and the dNTP supply in monocyte-derived macrophages (MDMs). RESULTS: IFN-α inhibited HIV-1 transduction in monocytes and in MDMs while SAMHD1 expression was not up-regulated. Vpx triggered SAMHD1 degradation in IFN-α treated cells, and weakly restored HIV-1 transduction from the IFN-α block. Vpx helper effect towards HIV-1 transduction was gradually inhibited with increasing doses of IFN-α. dNTP levels were not significantly affected in MDMs and CD4+ primary activated T lymphocytes by IFN-α and, in correlation with SAMHD1 degradation, restoration of dNTP levels by Vpx was efficient in MDMs treated with the cytokine. In contrast, IFN-α inhibited Vpx-mediated SAMHD1 degradation in THP-1 cells, where, accordingly, Vpx could not rescue HIV-1 transduction. CONCLUSION: Our results suggest that the early antiviral effect of IFN-α results from a mechanism independent of nucleotide depletion in MDMs. In addition, they indicate that the macrophage-like THP-1 cell line may provide a system to characterize an IFN-α-induced cell response that inhibits Vpx-mediated SAMHD1 degradation.


Asunto(s)
VIH-1/genética , Interferón-alfa/inmunología , Macrófagos/inmunología , Macrófagos/virología , Proteínas de Unión al GTP Monoméricas/inmunología , Transducción Genética , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Humanos , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleótidos/metabolismo , Proteolisis , Proteína 1 que Contiene Dominios SAM y HD , Proteínas Reguladoras y Accesorias Virales/metabolismo
8.
Front Vet Sci ; 10: 1235123, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37745217

RESUMEN

Since the first detection of rabbit hemorrhagic disease (RHD), the rabbit hemorrhagic disease virus (RHDV) has been responsible for high morbidity and mortality worldwide, both in domestic and in wild rabbits. Despite the apparent control of RHD in rabbitries through vaccination, several studies highlighted the rapid evolution of RHDV by recombination, which may facilitate the emergence of new pathogenic strains. The aim of this study was to confirm the presence and characterize RHDV in Algeria. For this, rabbit samples were collected in the north of Algeria, between 2018 and 2021, from small farms where the virus was suspected after the sudden death of a high number of rabbits, and from healthy hunted wild rabbits. The domestic rabbits revealed clinical signs and lesions that were suggestive of RHD. RT-PCR showed that 79.31% of the domestic rabbit samples were positive for RHDV, while in 20.69%, including the hunted rabbits, the virus was not detected. Phylogenetic analysis of the Algerian strains allowed the confirmation and identification as GI.2 (RHDV2), and showed a close relation to GI.3P-GI.2 recombinant strains, suggesting a potential introduction from other countries, with an older strain potentially originated from neighboring Tunisia, while more recent isolates grouped with strains from North America. Our study reports for the first time the presence of GI.2 (RHDV2) in Algeria with multiple routes of introduction. Consequently, we propose that RHDV control in Algeria should be based on epidemiological surveys in association with an adequate prophylactic program.

9.
J Biol Chem ; 286(27): 23742-52, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21566118

RESUMEN

Vpr, a small HIV auxiliary protein, hijacks the CUL4 ubiquitin ligase through DCAF1 to inactivate an unknown cellular target, leading to cell cycle arrest at the G(2) phase and cell death. Here we first sought to delineate the Vpr determinants involved in the binding to DCAF1 and to the target. On the one hand, the three α-helices of Vpr are necessary and sufficient for binding to DCAF1; on the other hand, nonlinear determinants in Vpr are required for binding to the target, as shown by using protein chimeras. We also underscore that a SRIG motif conserved in the C-terminal tail of Vpr proteins from HIV-1/SIVcpz and HIV-2/SIVsmm lineages is critical for G(2) arrest. Our results suggest that this motif may be predictive of the ability of Vpr proteins from other SIV lineages to mediate G(2) arrest. We took advantage of the characterization of a subset of G(2) arrest-defective, but DCAF1 binding-proficient mutants, to investigate whether Vpr interferes with cell viability independently of its ability to induce G(2) arrest. These mutants inhibited cell colony formation in HeLa cells and are cytotoxic in lymphocytes, unmasking a G(2) arrest-independent cytopathic effect of Vpr. Furthermore these mutants do not block cell cycle progression at the G(1) or S phases but trigger apoptosis through caspase 3. Disruption of DCAF1 binding restored efficiency of colony formation. However, DCAF1 binding per se is not sufficient to confer cytopathicity. These data support a model in which Vpr recruits DCAF1 to induce the degradation of two host proteins independently required for proper cell growth.


Asunto(s)
Proteínas Portadoras/metabolismo , Ciclo Celular , VIH-1/metabolismo , Modelos Biológicos , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Secuencias de Aminoácidos , Proteínas Portadoras/genética , Muerte Celular/genética , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Células HEK293 , VIH-1/genética , Células HeLa , Humanos , Mutación , Proteínas Serina-Treonina Quinasas , Ubiquitina-Proteína Ligasas , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/genética
10.
FASEB J ; 25(9): 3019-31, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21670065

RESUMEN

The physiological and pathological functions of proteinase 3 (PR3) are not well understood due to its close similarity to human neutrophil elastase (HNE) and the lack of a specific inhibitor. Based on structural analysis of the active sites of PR3 and HNE, we generated mutants derived from the polyvalent inhibitor SerpinB1 (monocyte/neutrophil elastase inhibitor) that specifically inhibit PR3 and that differ from wt-SerpinB1 by only 3 or 4 residues in the reactive center loop. The rate constant of association between the best SerpinB1 mutant and PR3 is 1.4 × 107 M⁻¹ · s⁻¹, which is ∼100-fold higher than that observed with wt-SerpinB1 and compares with that of α1-protease inhibitor (α1-PI) toward HNE. SerpinB1(S/DAR) is cleaved by HNE, but due to differences in rate, inhibition of PR3 by SerpinB1(S/DAR) is only minimally affected by the presence of HNE even when the latter is in excess. SerpinB1(S/DAR) inhibits soluble PR3 and also membrane-bound PR3 at the surface of activated neutrophils. Moreover, SerpinB1(S/DAR) clears induced PR3 from the surface of activated neutrophils. Overall, these specific inhibitors of PR3 will be valuable for defining biological functions of the protease and may prove useful as therapeutics for PR3-related inflammatory diseases, such as Wegener's granulomatosis.


Asunto(s)
Autoantígenos/metabolismo , Granulomatosis con Poliangitis/inmunología , Mieloblastina/antagonistas & inhibidores , Neutrófilos/efectos de los fármacos , Serpinas/farmacología , Autoanticuerpos/química , Autoanticuerpos/metabolismo , Clonación Molecular , Humanos , Modelos Moleculares , Mutación , Mieloblastina/metabolismo , Neutrófilos/metabolismo , Conformación Proteica , Proteínas Recombinantes , Serpinas/química
12.
Biochemistry ; 49(17): 3534-44, 2010 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-20359246

RESUMEN

The Mcmar1 mariner element (MLE) presents some intriguing features with two large, perfectly conserved, 355 bp inverted terminal repeats (ITRs) containing two 28 bp direct repeats (DRs). The presence of a complete ORF in Mcmar1 makes it possible to explore the transposition of this unusual MLE. Mcmar1 transposase (MCMAR1) was purified, and in vitro transposition assays showed that it is able to promote ITR-dependent DNA cleavages and recombination events, which correspond to plasmid fusions and transpositions with imprecise ends. Further analyses indicated that MCMAR1 is able to interact with the 355 bp ITR through two DRs: the EDR (external DR) is a high-affinity binding site for MCMAR1, whereas the IDR (internal DR) is a low-affinity binding site. The main complex detected within the EDR contained a transposase dimer and only one DNA molecule. We hypothesize that the inability of MCMAR1 to promote precise in vitro transposition events could be due to mutations in its ORF sequence or to the specific features of transposase binding to the ITR. Indeed, the ITR region spanning from EDR to IDR resembles a MITE and could be bent by specific host factors. This suggests that the assembly of the transposition complex is more complex than that of those involved in the mobility of the Mos1 and Himar1 mariner elements.


Asunto(s)
Elementos Transponibles de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Recombinación Genética , Secuencias Repetidas Terminales/genética , Transposasas/genética , Transposasas/metabolismo , Secuencia de Bases , Huella de ADN , Ensayo de Cambio de Movilidad Electroforética , Datos de Secuencia Molecular , Homología de Secuencia de Ácido Nucleico
13.
Mol Immunol ; 45(4): 1153-64, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17727952

RESUMEN

Mastitis is an inflammation of the mammary gland, most of the time caused by invading pathogens. Phagocytosis by neutrophils is a crucial defense of the mammary gland and the prompt recruitment of these phagocytes from blood to milk compartments is essential for the outcome of the infection. ELR+ CXC chemokines, ligands of the two interleukin-8 receptors (IL-8R), CXCR1 and CXCR2, are likely to be involved in the initiation of the inflammatory response and also in the migration of neutrophils. Recently, the polymorphism of bovine CXCR2 has been associated with resistance to mastitis. However, as the bovine IL-8R are not functionally defined, their contribution to the recruitment of neutrophils remains undetermined. In this study, the RNA ligase-mediated (RLM)-RACE method was used to clone a novel bovine interleukin-8 receptor (nIL-8R) of the bovine species. We showed that both bovine IL-8R (nIL-8R and the published CXCR2) are functional since bovine IL-8 induced migration of HEK-293 cells expressing either IL-8R. In addition, comparisons of full-length sequences suggested that the published CXCR2 sequence was improperly annotated and that the sequences of the nIL-8R and the published CXCR2 are homologous to human CXCR2 and CXCR1, respectively. This was confirmed by binding assays with labeled IL-8 and GRO-beta and calcium (Ca) flux responses of transfected cells. Moreover, the C-terminal of both bovine IL-8R showed 100% identity, whereas they differ in most other species, suggesting that the two bovine IL-8R initiate similar signal transduction. These results constitute a basis to improve our understanding of the molecular mechanisms implicated in the recruitment of bovine neutrophils.


Asunto(s)
Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Calcio/metabolismo , Bovinos , Línea Celular , Movimiento Celular , Quimiocina CXCL2/metabolismo , Humanos , Interleucina-8/metabolismo , Datos de Secuencia Molecular , Ensayo de Unión Radioligante , Homología de Secuencia de Aminoácido , Transducción de Señal , Especificidad de la Especie
14.
Nat Microbiol ; 3(8): 891-897, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29891865

RESUMEN

To evade host immune defences, human immunodeficiency viruses 1 and 2 (HIV-1 and HIV-2) have evolved auxiliary proteins that target cell restriction factors. Viral protein X (Vpx) from the HIV-2/SIVsmm lineage enhances viral infection by antagonizing SAMHD1 (refs 1,2), but this antagonism is not sufficient to explain all Vpx phenotypes. Here, through a proteomic screen, we identified another Vpx target-HUSH (TASOR, MPP8 and periphilin)-a complex involved in position-effect variegation3. HUSH downregulation by Vpx is observed in primary cells and HIV-2-infected cells. Vpx binds HUSH and induces its proteasomal degradation through the recruitment of the DCAF1 ubiquitin ligase adaptor, independently from SAMHD1 antagonism. As a consequence, Vpx is able to reactivate HIV latent proviruses, unlike Vpx mutants, which are unable to induce HUSH degradation. Although antagonism of human HUSH is not conserved among all lentiviral lineages including HIV-1, it is a feature of viral protein R (Vpr) from simian immunodeficiency viruses (SIVs) of African green monkeys and from the divergent SIV of l'Hoest's monkey, arguing in favour of an ancient lentiviral species-specific vpx/vpr gene function. Altogether, our results suggest the HUSH complex as a restriction factor, active in primary CD4+ T cells and counteracted by Vpx, therefore providing a molecular link between intrinsic immunity and epigenetic control.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Lentivirus de los Primates/fisiología , Proteínas Nucleares/metabolismo , Fosfoproteínas/metabolismo , Proteómica/métodos , Proteínas Reguladoras y Accesorias Virales/metabolismo , Línea Celular , Regulación hacia Abajo , Regulación de la Expresión Génica , Células HEK293 , VIH-2/metabolismo , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Células Jurkat , Lentivirus de los Primates/metabolismo , Provirus/metabolismo , Virus de la Inmunodeficiencia de los Simios/metabolismo , Células THP-1
16.
PLoS One ; 8(10): e77320, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24116224

RESUMEN

The Vpr protein from type 1 and type 2 Human Immunodeficiency Viruses (HIV-1 and HIV-2) is thought to inactivate several host proteins through the hijacking of the DCAF1 adaptor of the Cul4A ubiquitin ligase. Here, we identified two transcriptional regulators, ZIP and sZIP, as Vpr-binding proteins degraded in the presence of Vpr. ZIP and sZIP have been shown to act through the recruitment of the NuRD chromatin remodeling complex. Strikingly, chromatin is the only cellular fraction where Vpr is present together with Cul4A ubiquitin ligase subunits. Components of the NuRD complex and exogenous ZIP and sZIP were also associated with this fraction. Several lines of evidence indicate that Vpr induces ZIP and sZIP degradation by hijacking DCAF1: (i) Vpr induced a drastic decrease of exogenously expressed ZIP and sZIP in a dose-dependent manner, (ii) this decrease relied on the proteasome activity, (iii) ZIP or sZIP degradation was impaired in the presence of a DCAF1-binding deficient Vpr mutant or when DCAF1 expression was silenced. Vpr-mediated ZIP and sZIP degradation did not correlate with the growth-related Vpr activities, namely G2 arrest and G2 arrest-independent cytotoxicity. Nonetheless, infection with HIV-1 viruses expressing Vpr led to the degradation of the two proteins. Altogether our results highlight the existence of two host transcription factors inactivated by Vpr. The role of Vpr-mediated ZIP and sZIP degradation in the HIV-1 replication cycle remains to be deciphered.


Asunto(s)
Proteínas Portadoras/metabolismo , Infecciones por VIH/metabolismo , VIH-1/fisiología , Interacciones Huésped-Patógeno , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Proteínas Represoras/metabolismo , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Ensamble y Desensamble de Cromatina , Células HEK293 , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas , Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo
17.
Cytokine ; 38(1): 12-21, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17532224

RESUMEN

We studied the inflammatory and immune responses of bovine mammary epithelial cells (bMEC) infected by mastitis isolates of Staphylococcus aureus. Primary cultures of bMEC were co-incubated separately with three strains of S. aureus and one strain of Escherichia coli. Transcriptional levels and/or protein release of interleukin-8 (IL-8), growth related oncogene alpha (GRO-alpha), growth related oncogene beta (GRO-beta), tumor necrosis factor alpha (TNF-alpha), interleukin-1beta (IL-1beta), transforming growth factor beta1 (TGF-beta1) and interleukin-10 (IL-10) were measured at 3, 10 and 24h post-infection (PI). The results indicated that at earlier hours of co-culture, bMEC infected with S. aureus or E. coli expressed more IL-1beta, TNF-alpha, IL-8 and GRO-alpha mRNA than uninfected bMEC. Furthermore, infected bMEC released more TNF-alpha, IL-8, GRO-alpha and GRO-beta proteins than uninfected bMEC. However, differential transcription and release of some cytokines/chemokines from bMEC was observed according to the strain of S. aureus and bacteria Gram type. In conclusion, bMEC did not show an anti-inflammatory potential through IL-10 or TGF-beta1 release. Nevertheless, bMEC were able to release neutrophil-mobilizing chemokines and pro-inflammatory cytokines upon bacterial stimulation, strongly suggesting that bMEC are active contributors to immune and inflammatory responses of mammary gland. In addition, the clinical characteristics and resolution of mastitis may be partly determined by the responses of bMEC according to S. aureus strains and bacteria Gram type.


Asunto(s)
Quimiocinas/metabolismo , Citocinas/metabolismo , Escherichia coli/inmunología , Glándulas Mamarias Animales/inmunología , Staphylococcus aureus/inmunología , Animales , Bovinos , Forma de la Célula , Células Cultivadas , Quimiocinas/análisis , Quimiocinas/genética , Citocinas/análisis , Citocinas/genética , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Células Epiteliales/microbiología , Glándulas Mamarias Animales/microbiología , Neutrófilos/citología , ARN Mensajero/análisis , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA